Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Microbiol ; 7(5): 707-715, 2022 05.
Article in English | MEDLINE | ID: mdl-35437328

ABSTRACT

The mosquito microbiota can influence host physiology and vector competence, but a detailed understanding of these processes is lacking. Here we found that the gut microbiota of Anopheles stephensi, a competent malaria vector, is involved in tryptophan metabolism and is responsible for the catabolism of the peritrophic matrix impairing tryptophan metabolites. Antibiotic elimination of the microbiota led to the accumulation of tryptophan and its metabolites-kynurenine, 3-hydroxykynurenine (3-HK) and xanthurenic acid. Of these metabolites, 3-HK impaired the structure of the peritrophic matrix and promoted Plasmodium berghei infection. Among the major gut microbiota members in A. stephensi, Pseudomonas alcaligenes catabolized 3-HK as revealed by whole-genome sequencing and LC-MS metabolic analysis. The genome of P. alcaligenes encodes kynureninase (KynU) that is responsible for the conversion of 3-HK to 3-hydroxyanthranilic acid. Mutation of KynU resulted in a P. alcaligenes strain that was unable to metabolize 3-HK and unable to protect the peritrophic matrix. Colonization of A. stephensi with KynU-mutated P. alcaligenes failed to protect mosquitoes against parasite infection as compared with mosquitoes colonized with wild-type P. alcaligenes. In summary, this study identifies an unexpected function of mosquito gut microbiota in controlling mosquito tryptophan metabolism, with important implications for vector competence.


Subject(s)
Anopheles , Gastrointestinal Microbiome , Malaria , Animals , Anopheles/parasitology , Malaria/parasitology , Mosquito Vectors/genetics , Tryptophan
2.
J Am Mosq Control Assoc ; 37(1): 38-40, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33857314

ABSTRACT

Microbiota are vital for the development, physiology, and vectorial capacity of mosquitoes. The composition and role of microbiota in Anopheles species, especially Anopheles gambiae and Anopheles stephensi, have been extensively studied, but little is known about the microbiota of Anopheles species in China. We characterized the microbial communities of Anopheles dirus, Anopheles sinensis, and Anopheles lesteri by 16S rRNA sequencing. There were distinct differences in the composition of microbiota in An. lesteri and the other 2 species. The discriminatory genera in the 3 species were analyzed by the linear discriminant analysis effect size method. Our results provide an overview of the population structure of microbiota in 3 native Anopheles species and will pave the way for further understanding of their role in mosquito physiology and vector competence.


Subject(s)
Anopheles/microbiology , Bacteria/isolation & purification , Microbiota , Mosquito Vectors/microbiology , Animals , China , RNA, Bacterial/analysis , RNA, Ribosomal, 16S/analysis , Species Specificity
3.
Cell Rep ; 35(3): 108992, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882310

ABSTRACT

Plant-nectar-derived sugar is the major energy source for mosquitoes, but its influence on vector competence for malaria parasites remains unclear. Here, we show that Plasmodium berghei infection of Anopheles stephensi results in global metabolome changes, with the most significant impact on glucose metabolism. Feeding on glucose or trehalose (the main hemolymph sugars) renders the mosquito more susceptible to Plasmodium infection by alkalizing the mosquito midgut. The glucose/trehalose diets promote proliferation of a commensal bacterium, Asaia bogorensis, that remodels glucose metabolism in a way that increases midgut pH, thereby promoting Plasmodium gametogenesis. We also demonstrate that the sugar composition from different natural plant nectars influences A. bogorensis growth, resulting in a greater permissiveness to Plasmodium. Altogether, our results demonstrate that dietary glucose is an important determinant of mosquito vector competency for Plasmodium, further highlighting a key role for mosquito-microbiota interactions in regulating the development of the malaria parasite.


Subject(s)
Acetobacteraceae/metabolism , Anopheles/metabolism , Glucose/pharmacology , Metabolome , Mosquito Vectors/metabolism , Trehalose/pharmacology , Acetobacteraceae/growth & development , Animals , Anopheles/drug effects , Anopheles/microbiology , Anopheles/parasitology , Digestive System/microbiology , Digestive System/parasitology , Female , Gametogenesis/drug effects , Gametogenesis/genetics , Gene Expression Regulation , Glucose/metabolism , Host-Pathogen Interactions/genetics , Hydrogen-Ion Concentration , Life Cycle Stages/drug effects , Life Cycle Stages/genetics , Malaria/parasitology , Microbiota/genetics , Mosquito Vectors/drug effects , Mosquito Vectors/microbiology , Mosquito Vectors/parasitology , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Symbiosis/genetics , Trehalose/metabolism
4.
PLoS Pathog ; 17(2): e1009353, 2021 02.
Article in English | MEDLINE | ID: mdl-33626094

ABSTRACT

Repeated blood meals provide essential nutrients for mosquito egg development and routes for pathogen transmission. The target of rapamycin, the TOR pathway, is essential for vitellogenesis. However, its influence on pathogen transmission remains to be elucidated. Here, we show that rapamycin, an inhibitor of the TOR pathway, effectively suppresses Plasmodium berghei infection in Anopheles stephensi. An. stephensi injected with rapamycin or feeding on rapamycin-treated mice showed increased resistance to P. berghei infection. Exposing An. stephensi to a rapamycin-coated surface not only decreased the numbers of both oocysts and sporozoites but also impaired mosquito survival and fecundity. Transcriptome analysis revealed that the inhibitory effect of rapamycin on parasite infection was through the enhanced activation of immune responses, especially the NF-κB transcription factor REL2, a regulator of the immune pathway and complement system. Knockdown of REL2 in rapamycin-treated mosquitoes abrogated the induction of the complement-like proteins TEP1 and SPCLIP1 and abolished rapamycin-mediated refractoriness to Plasmodium infection. Together, these findings demonstrate a key role of the TOR pathway in regulating mosquito immune responses, thereby influencing vector competence.


Subject(s)
Anopheles/drug effects , Immunity, Innate/immunology , Malaria/drug therapy , Mosquito Vectors/drug effects , Plasmodium berghei/pathogenicity , Sirolimus/pharmacology , Animals , Anopheles/immunology , Anopheles/parasitology , Female , Gene Expression Profiling , Immunity, Innate/drug effects , Immunosuppressive Agents/pharmacology , Malaria/immunology , Malaria/parasitology , Malaria/transmission , Mice , Mice, Inbred BALB C , Mosquito Vectors/immunology , Mosquito Vectors/parasitology , Oocysts/drug effects , Oocysts/growth & development , Oocysts/immunology , Sporozoites/drug effects , Sporozoites/growth & development , Sporozoites/immunology
5.
Korean J Parasitol ; 56(5): 515-519, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30419739

ABSTRACT

Triatoma rubrofasciata is a wide-spread vector of Chagas disease in Americas. In this study, we completed the mitochondrial genome sequencing of T. rubrofasciata. The total length of T. rubrofasciata mitochondrial genome was 17,150 bp with the base composition of 40.4% A, 11.6% G, 29.4% T and 18.6% C. It included 13 protein-coding genes, 22 tRNA genes, 2 rRNA genes and one control region. We constructed a phylogenetic tree on the 13 protein-coding genes of T. rubrofasciata and other 13 closely related species to show their phylogenic relationship. The determination of T. rubrofasciata mitogenome would play an important role in understanding the genetic diversity and evolution of triatomine bugs.


Subject(s)
Chagas Disease , Disease Vectors , Genome, Insect/genetics , Genome, Mitochondrial/genetics , Triatoma/genetics , Whole Genome Sequencing , Animals , Chagas Disease/parasitology , Chagas Disease/transmission , Genetic Variation , Humans , Open Reading Frames/genetics , Phylogeny , RNA, Ribosomal , RNA, Transfer
SELECTION OF CITATIONS
SEARCH DETAIL
...